38 results on '"Ravi VM"'
Search Results
2. Spatial T-cell receptor sequencing (SPTCR-seq) uncovers regional anti-tumour immunity in glioblastoma
- Author
-
Kada Benotmane, J, Kückelhaus, J, Will, P, Schnell, O, Beck, J, Ravi, VM, Joseph, K, and Heiland, DH
- Subjects
ddc: 610 ,Medicine and health ,natural sciences - Abstract
Objective: An understanding of the diversity of T cell response and clonality in the inherently heterogeneous glioblastoma (GBM) is of paramount importance to explore underlying mechanisms of anti-tumor immunity. Here, we present SPatial T-Cell Receptor sequencing (SPTCR-seq), a novel method to integrate [for full text, please go to the a.m. URL]
- Published
- 2022
- Full Text
- View/download PDF
3. Personalised therapeutic approach in translational neuro-oncology - JAK/STAT inhibition reverses the anti-tumour immunity induced by myeloid cells in T cells
- Author
-
Ravi, VM, Neidert, N, Joseph, K, Beck, J, Schnell, O, Heiland, DH, Ravi, VM, Neidert, N, Joseph, K, Beck, J, Schnell, O, and Heiland, DH
- Published
- 2022
4. The role of onco-metabolite (R-2-hydroxyglutarate) in the IDH mutant glioblastoma microenvironment
- Author
-
Ravi, VM, Behringer, S, Joseph, K, Beck, J, Schnell, O, Heiland, DH, Ravi, VM, Behringer, S, Joseph, K, Beck, J, Schnell, O, and Heiland, DH
- Published
- 2022
5. Metabotropic glutamate receptor III inhibition sensitises GBM to chemotherapeutics
- Author
-
Joseph, K, Maier, J, Ravi, VM, Beck, J, Hofmann, U, Heiland, DH, Schnell, O, Joseph, K, Maier, J, Ravi, VM, Beck, J, Hofmann, U, Heiland, DH, and Schnell, O
- Published
- 2022
6. A target therapy approach for BRAF mutant Glioblastoma with human preclinical glioblastoma model
- Author
-
Zhang, J, Ravi, VM, Joseph, K, Beck, J, Schnell, O, Heiland, DH, Zhang, J, Ravi, VM, Joseph, K, Beck, J, Schnell, O, and Heiland, DH
- Published
- 2022
7. Lineage tracking of glioma-associated myeloid cells by high-dimensional techniques
- Author
-
Neidert, N, Joseph, K, Ravi, VM, Behringer, SP, Maier, JP, Beck, J, Schnell, O, and Heiland, DH
- Subjects
ddc: 610 ,610 Medical sciences ,Medicine - Abstract
Objective: In malignant brain tumours, crosstalk and adaptation of brain resident and infiltrating myeloid cells form an immunosuppressive environment that causes dysfunctional activation of the T cells and leads to an inadequate immune response. Most recently, checkpoint inhibition (PD-1) failed to[for full text, please go to the a.m. URL], 71. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC), 9. Joint Meeting mit der Japanischen Gesellschaft für Neurochirurgie
- Published
- 2020
- Full Text
- View/download PDF
8. Exploiting autocrine glutamate signalling via mGluR3 for temozolomide sensitisation
- Author
-
Maier, JP, Joseph, K, Behringer, SP, Ravi, VM, Neidert, N, Beck, J, Schnell, O, and Heiland, DH
- Subjects
ddc: 610 ,610 Medical sciences ,Medicine - Abstract
Objective: Invariably, malignant glioma develops resistance against temozolomide (TMZ) chemotherapy during the course of the disease through clonal selection of resistant cells. Autocrine glutamate signalling via metabotropic glutamate receptors (mGluR) was reported to sustain malignant hallmarks, however[for full text, please go to the a.m. URL], 71. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC), 9. Joint Meeting mit der Japanischen Gesellschaft für Neurochirurgie
- Published
- 2020
- Full Text
- View/download PDF
9. Multiomic mapping of spatiotemporal architecture identified decoupled lineage and reactive transcriptional states in glioblastoma
- Author
-
Ravi, VM, Will, P, Beck, J, Hofmann, U, Schnell, O, Heiland, DH, Ravi, VM, Will, P, Beck, J, Hofmann, U, Schnell, O, and Heiland, DH
- Published
- 2021
10. Environmental effect on microglial cell depletion in organotypic brain slice cultures
- Author
-
Ravi, VM, Joseph, K, Beck, J, Hofmann, U, Schnell, O, Heiland, DH, Ravi, VM, Joseph, K, Beck, J, Hofmann, U, Schnell, O, and Heiland, DH
- Published
- 2021
11. The role of aging induced environmental alterations in glioblastoma
- Author
-
Ravi, VM, Will, P, Beck, J, Hofmann, U, Schnell, O, Heiland, DH, Ravi, VM, Will, P, Beck, J, Hofmann, U, Schnell, O, and Heiland, DH
- Published
- 2021
12. Personalised treatment option in translational neuro-oncology - JAK/STAT-inhibition reverses myeloid cell-induced anti-tumour-immunity in T cells
- Author
-
Heiland, DH, Ravi, VM, Neidert, N, Hofmann, U, Beck, J, Schnell, O, Heiland, DH, Ravi, VM, Neidert, N, Hofmann, U, Beck, J, and Schnell, O
- Published
- 2021
13. Spatiotemporal diversity of cell communication in glioblastoma aid transcriptional plasticity
- Author
-
Joseph, K, Vollmer, L, Ravi, VM, Beck, J, Hofmann, U, Schnell, O, Heiland, DH, Joseph, K, Vollmer, L, Ravi, VM, Beck, J, Hofmann, U, Schnell, O, and Heiland, DH
- Published
- 2021
14. Reshaping the GBM landscape to enable targeting of chemo-resistant tumours
- Author
-
Joseph, K, Maier, J, Ravi, VM, Beck, J, Hofmann, U, Schnell, O, Heiland, DH, Joseph, K, Maier, J, Ravi, VM, Beck, J, Hofmann, U, Schnell, O, and Heiland, DH
- Published
- 2021
15. Meclofenamate demolishes the network architecture and electrophysiological coupling of malignant gliomas
- Author
-
Schneider, M, Potthoff, AL, Vollmer, L, Ravi, VM, Evert, BO, Güresir, E, Schuss, P, Dolf, A, Westhoff, MA, Beck, J, Vatter, H, Waha, A, Schnell, O, Herrlinger, U, Heiland, DH, Schneider, M, Potthoff, AL, Vollmer, L, Ravi, VM, Evert, BO, Güresir, E, Schuss, P, Dolf, A, Westhoff, MA, Beck, J, Vatter, H, Waha, A, Schnell, O, Herrlinger, U, and Heiland, DH
- Published
- 2021
16. A novel glioblastoma invasion model using human brain slice cultures
- Author
-
Ravi, VM, Joseph, K, Neidert, N, Beck, J, Hofmann, UG, Schnell, O, Heiland, DH, Ravi, VM, Joseph, K, Neidert, N, Beck, J, Hofmann, UG, Schnell, O, and Heiland, DH
- Published
- 2020
17. Mapping the developmental trajectory of human astrocytes reveals divergence in glioblastoma.
- Author
-
Sojka C, Wang HV, Bhatia TN, Li Y, Chopra P, Sing A, Voss A, King A, Wang F, Joseph K, Ravi VM, Olson J, Hoang K, Nduom E, Corces VG, Yao B, and Sloan SA
- Abstract
Glioblastoma (GBM) is defined by heterogeneous and resilient cell populations that closely reflect neurodevelopmental cell types. Although it is clear that GBM echoes early and immature cell states, identifying the specific developmental programmes disrupted in these tumours has been hindered by a lack of high-resolution trajectories of glial and neuronal lineages. Here we delineate the course of human astrocyte maturation to uncover discrete developmental stages and attributes mirrored by GBM. We generated a transcriptomic and epigenomic map of human astrocyte maturation using cortical organoids maintained in culture for nearly 2 years. Through this approach, we chronicled a multiphase developmental process. Our time course of human astrocyte maturation includes a molecularly distinct intermediate period that serves as a lineage commitment checkpoint upstream of mature quiescence. This intermediate stage acts as a site of developmental deviation separating IDH-wild-type neoplastic astrocyte-lineage cells from quiescent astrocyte populations. Interestingly, IDH1-mutant tumour astrocyte-lineage cells are the exception to this developmental perturbation, where immature properties are suppressed as a result of D-2-hydroxyglutarate oncometabolite exposure. We propose that this defiance is a consequence of IDH1-mutant-associated epigenetic dysregulation, and we identified biased DNA hydroxymethylation (5hmC) in maturation genes as a possible mechanism. Together, this study illustrates a distinct cellular state aberration in GBM astrocyte-lineage cells and presents developmental targets for experimental and therapeutic exploration., Competing Interests: Competing interests: The authors declare no competing interests. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript., (© 2025. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2025
- Full Text
- View/download PDF
18. ZBTB18 regulates cytokine expression and affects microglia/macrophage recruitment and commitment in glioblastoma.
- Author
-
Ferrarese R, Joseph K, Andrieux G, Haase IV, Zanon F, Kling E, Izzo A, Corrales E, Schwabenland M, Prinz M, Ravi VM, Boerries M, Heiland DH, and Carro MS
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Macrophages metabolism, Macrophages immunology, Gene Expression Regulation, Neoplastic, Brain Neoplasms genetics, Brain Neoplasms pathology, Brain Neoplasms metabolism, Brain Neoplasms immunology, Cell Movement genetics, Glioblastoma genetics, Glioblastoma pathology, Glioblastoma metabolism, Microglia metabolism, Cytokines metabolism, Repressor Proteins genetics, Repressor Proteins metabolism
- Abstract
Glioma associated macrophages/microglia (GAMs) play an important role in glioblastoma (GBM) progression, due to their massive recruitment to the tumor site and polarization to a tumor promoting phenotype. GAMs secrete a variety of cytokines, which facilitate tumor cell growth and invasion, and prevent other immune cells from mounting an immune response against the tumor. Here, we demonstrate that zinc finger and BTB containing domain 18 (ZBTB18), a transcriptional repressor with tumor suppressive function in glioblastoma, impairs the production of key cytokines, which function as chemoattractant for GAMs. Consistently, we observe a reduced migration of GAMs when ZBTB18 is expressed by glioblastoma cells, both in cell culture and in vivo experiments. Moreover, RNA sequencing analysis shows that the presence of ZBTB18 in glioblastoma cells alters the commitment of conditioned microglia, suggesting the loss of the immune-suppressive phenotype and the acquisition of pro-inflammatory features. Thus, therapeutic approaches to increase ZBTB18 expression in GBM cells could represent an effective adjuvant to immune therapy in GBM., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
19. SMAC mimetic drives microglia phenotype and glioblastoma immune microenvironment.
- Author
-
Snacel-Fazy E, Soubéran A, Grange M, Joseph K, Colin C, Morando P, Luche H, Pagano A, Brustlein S, Debarbieux F, Toutain S, Siret C, van de Pavert SA, Rougon G, Figarella-Branger D, Ravi VM, Tabouret E, and Tchoghandjian A
- Subjects
- Animals, Humans, Mice, Brain Neoplasms immunology, Brain Neoplasms pathology, Apoptosis Regulatory Proteins metabolism, Mice, Inbred C57BL, Mitochondrial Proteins metabolism, Cell Line, Tumor, Tumor-Associated Macrophages immunology, Tumor-Associated Macrophages metabolism, Tumor-Associated Macrophages drug effects, Intracellular Signaling Peptides and Proteins metabolism, Mice, Transgenic, Glioblastoma immunology, Glioblastoma pathology, Tumor Microenvironment drug effects, Tumor Microenvironment immunology, Microglia drug effects, Microglia metabolism, Microglia immunology, Phenotype
- Abstract
Tumor-associated macrophages/microglia (TAMs) are highly plastic and heterogeneous immune cells that can be immune-supportive or tumor-supportive depending of the microenvironment. TAMs are the most abundant immune cells in glioblastoma (GB), and play a key role in immunosuppression. Therefore, TAMs reprogramming toward immune-supportive cells is a promising strategy to overcome immunosuppression. By leveraging scRNAseq human GB databases, we identified that Inhibitor of Apoptosis Proteins (IAP) were expressed by TAMs. To investigate their role in TAMs-related immunosuppression, we antagonized IAP using the central nervous system permeant SMAC mimetic GDC-0152 (SMg). On explants and cultured immune cells isolated from human GB samples, SMg modified TAMs activity. We showed that SMg treatment promoted microglia pro-apoptotic and anti-tumoral function via caspase-3 pro-inflammatory cleavage and the inhibition of tumoroids growth. Then we designed a relevant immunogenic mouse GB model to decipher the spatio-temporal densities, distribution, phenotypes and function of TAMs with or without SMg treatment. We used 3D imaging techniques, a transgenic mouse with fluorescent TAM subsets and mass cytometry. We confirmed that SMg promoted microglia activation, antigen-presenting function and tumor infiltration. In addition, we observed a remodeling of blood vessels, a decrease in anti-inflammatory macrophages and an increased level of monocytes and their mo-DC progeny. This remodeling of the TAM landscape is associated with an increase in CD8 T cell density and activation. Altogether, these results demonstrated that SMg drives the immunosuppressive basal microglia toward an active phenotype with pro-apoptotic and anti-tumoral function and modifies the GB immune landscape. This identifies IAP as targets of choice for a potential mechanism-based therapeutic strategy and SMg as a promising molecule for this application., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
20. Editorial: Spatiotemporal heterogeneity in CNS tumors.
- Author
-
Faisal SM, Ravi VM, and Miska JM
- Subjects
- Humans, Animals, Tumor Microenvironment immunology, Central Nervous System Neoplasms
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.
- Published
- 2024
- Full Text
- View/download PDF
21. VC-resist glioblastoma cell state: vessel co-option as a key driver of chemoradiation resistance.
- Author
-
Pichol-Thievend C, Anezo O, Pettiwala AM, Bourmeau G, Montagne R, Lyne AM, Guichet PO, Deshors P, Ballestín A, Blanchard B, Reveilles J, Ravi VM, Joseph K, Heiland DH, Julien B, Leboucher S, Besse L, Legoix P, Dingli F, Liva S, Loew D, Giani E, Ribecco V, Furumaya C, Marcos-Kovandzic L, Masliantsev K, Daubon T, Wang L, Diaz AA, Schnell O, Beck J, Servant N, Karayan-Tapon L, Cavalli FMG, and Seano G
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Chemoradiotherapy methods, Drug Resistance, Neoplasm, Gene Expression Regulation, Neoplastic, Radiation Tolerance, YAP-Signaling Proteins metabolism, Brain metabolism, Brain pathology, Proteomics, Glioblastoma metabolism, Glioblastoma pathology, Glioblastoma drug therapy, Glioblastoma genetics, Brain Neoplasms metabolism, Brain Neoplasms pathology, Brain Neoplasms drug therapy, Brain Neoplasms genetics
- Abstract
Glioblastoma (GBM) is a highly lethal type of cancer. GBM recurrence following chemoradiation is typically attributed to the regrowth of invasive and resistant cells. Therefore, there is a pressing need to gain a deeper understanding of the mechanisms underlying GBM resistance to chemoradiation and its ability to infiltrate. Using a combination of transcriptomic, proteomic, and phosphoproteomic analyses, longitudinal imaging, organotypic cultures, functional assays, animal studies, and clinical data analyses, we demonstrate that chemoradiation and brain vasculature induce cell transition to a functional state named VC-Resist (vessel co-opting and resistant cell state). This cell state is midway along the transcriptomic axis between proneural and mesenchymal GBM cells and is closer to the AC/MES1-like state. VC-Resist GBM cells are highly vessel co-opting, allowing significant infiltration into the surrounding brain tissue and homing to the perivascular niche, which in turn induces even more VC-Resist transition. The molecular and functional characteristics of this FGFR1-YAP1-dependent GBM cell state, including resistance to DNA damage, enrichment in the G2M phase, and induction of senescence/stemness pathways, contribute to its enhanced resistance to chemoradiation. These findings demonstrate how vessel co-option, perivascular niche, and GBM cell plasticity jointly drive resistance to therapy during GBM recurrence., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
22. High-sensitive spatially resolved T cell receptor sequencing with SPTCR-seq.
- Author
-
Benotmane JK, Kueckelhaus J, Will P, Zhang J, Ravi VM, Joseph K, Sankowski R, Beck J, Lee-Chang C, Schnell O, and Heiland DH
- Subjects
- Complementarity Determining Regions genetics, High-Throughput Nucleotide Sequencing methods, Gene Expression Profiling, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell genetics, T-Lymphocytes
- Abstract
Spatial resolution of the T cell repertoire is essential for deciphering cancer-associated immune dysfunction. Current spatially resolved transcriptomic technologies are unable to directly annotate T cell receptors (TCR). We present spatially resolved T cell receptor sequencing (SPTCR-seq), which integrates optimized target enrichment and long-read sequencing for highly sensitive TCR sequencing. The SPTCR computational pipeline achieves yield and coverage per TCR comparable to alternative single-cell TCR technologies. Our comparison of PCR-based and SPTCR-seq methods underscores SPTCR-seq's superior ability to reconstruct the entire TCR architecture, including V, D, J regions and the complementarity-determining region 3 (CDR3). Employing SPTCR-seq, we assess local T cell diversity and clonal expansion across spatially discrete niches. Exploration of the reciprocal interaction of the tumor microenvironmental and T cells discloses the critical involvement of NK and B cells in T cell exhaustion. Integrating spatially resolved omics and TCR sequencing provides as a robust tool for exploring T cell dysfunction in cancers and beyond., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
23. Isolation and profiling of viable tumor cells from human ex vivo glioblastoma cultures through single-cell transcriptomics.
- Author
-
Zhang J, Straehle J, Joseph K, Neidert N, Behringer S, Göldner J, Vlachos A, Prinz M, Fung C, Beck J, Schnell O, Heiland DH, and Ravi VM
- Subjects
- Humans, Gene Expression Profiling, Brain, Transcriptome genetics, Glioblastoma genetics, Brain Neoplasms genetics
- Abstract
Single-cell RNA-sequencing (scRNA-seq) is becoming a ubiquitous method in profiling the cellular transcriptomes of both malignant and non-malignant cells from the human brain. Here, we present a protocol to isolate viable tumor cells from human ex vivo glioblastoma cultures for single-cell transcriptomic analysis. We describe steps including surgical tissue collection, sectioning, culturing, primary tumor cells inoculation, growth tracking, fluorescence-based cell sorting, and population-enriched scRNA-seq. This comprehensive methodology empowers in-depth understanding of brain tumor biology at the single-cell level. For complete details on the use and execution of this protocol, please refer to Ravi et al.
1 ., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
24. Technical report: surgical preparation of human brain tissue for clinical and basic research.
- Author
-
Straehle J, Ravi VM, Heiland DH, Galanis C, Lenz M, Zhang J, Neidert NN, El Rahal A, Vasilikos I, Kellmeyer P, Scheiwe C, Klingler JH, Fung C, Vlachos A, Beck J, and Schnell O
- Subjects
- Humans, Neurosurgical Procedures methods, Microdissection, Preoperative Care, Brain surgery, Brain Neoplasms surgery
- Abstract
Background: The study of the distinct structure and function of the human central nervous system, both in healthy and diseased states, is becoming increasingly significant in the field of neuroscience. Typically, cortical and subcortical tissue is discarded during surgeries for tumors and epilepsy. Yet, there is a strong encouragement to utilize this tissue for clinical and basic research in humans. Here, we describe the technical aspects of the microdissection and immediate handling of viable human cortical access tissue for basic and clinical research, highlighting the measures needed to be taken in the operating room to ensure standardized procedures and optimal experimental results., Methods: In multiple rounds of experiments (n = 36), we developed and refined surgical principles for the removal of cortical access tissue. The specimens were immediately immersed in cold carbogenated N-methyl-D-glucamine-based artificial cerebrospinal fluid for electrophysiology and electron microscopy experiments or specialized hibernation medium for organotypic slice cultures., Results: The surgical principles of brain tissue microdissection were (1) rapid preparation (<1 min), (2) maintenance of the cortical axis, (3) minimization of mechanical trauma to sample, (4) use of pointed scalpel blade, (5) avoidance of cauterization and blunt preparation, (6) constant irrigation, and (7) retrieval of the sample without the use of forceps or suction. After a single round of introduction to these principles, multiple surgeons adopted the technique for samples with a minimal dimension of 5 mm spanning all cortical layers and subcortical white matter. Small samples (5-7 mm) were ideal for acute slice preparation and electrophysiology. No adverse events from sample resection were observed., Conclusion: The microdissection technique of human cortical access tissue is safe and easily adoptable into the routine of neurosurgical procedures. The standardized and reliable surgical extraction of human brain tissue lays the foundation for human-to-human translational research on human brain tissue., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
25. Quantitative proteomic landscapes of primary and recurrent glioblastoma reveal a protumorigeneic role for FBXO2-dependent glioma-microenvironment interactions.
- Author
-
Buehler M, Yi X, Ge W, Blattmann P, Rushing E, Reifenberger G, Felsberg J, Yeh C, Corn JE, Regli L, Zhang J, Cloos A, Ravi VM, Wiestler B, Heiland DH, Aebersold R, Weller M, Guo T, and Weiss T
- Subjects
- Humans, Animals, Mice, Proteomics, Mice, Knockout, Brain pathology, Proteins, Tumor Microenvironment, Neoplasm Proteins, Nerve Tissue Proteins, Cell Cycle Proteins, Glioblastoma pathology, Glioma pathology, Brain Neoplasms pathology, F-Box Proteins genetics
- Abstract
Background: Recent efforts have described the evolution of glioblastoma from initial diagnosis to post-treatment recurrence on a genomic and transcriptomic level. However, the evolution of the proteomic landscape is largely unknown., Methods: Sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) was used to characterize the quantitative proteomes of two independent cohorts of paired newly diagnosed and recurrent glioblastomas. Recurrence-associated proteins were validated using immunohistochemistry and further studied in human glioma cell lines, orthotopic xenograft models, and human organotypic brain slice cultures. External spatial transcriptomic, single-cell, and bulk RNA sequencing data were analyzed to gain mechanistic insights., Results: Although overall proteomic changes were heterogeneous across patients, we identified BCAS1, INF2, and FBXO2 as consistently upregulated proteins at recurrence and validated these using immunohistochemistry. Knockout of FBXO2 in human glioma cells conferred a strong survival benefit in orthotopic xenograft mouse models and reduced invasive growth in organotypic brain slice cultures. In glioblastoma patient samples, FBXO2 expression was enriched in the tumor infiltration zone and FBXO2-positive cancer cells were associated with synaptic signaling processes., Conclusions: These findings demonstrate a potential role of FBXO2-dependent glioma-microenvironment interactions to promote tumor growth. Furthermore, the published datasets provide a valuable resource for further studies., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
26. 'Hippocampal innate inflammatory gliosis only' in pharmacoresistant temporal lobe epilepsy.
- Author
-
Grote A, Heiland DH, Taube J, Helmstaedter C, Ravi VM, Will P, Hattingen E, Schüre JR, Witt JA, Reimers A, Elger C, Schramm J, Becker AJ, and Delev D
- Subjects
- Humans, Gliosis pathology, Sclerosis pathology, Hippocampus pathology, Temporal Lobe pathology, Treatment Outcome, Epilepsy, Temporal Lobe pathology, Drug Resistant Epilepsy complications, Hippocampal Sclerosis
- Abstract
Drug-resistant mesial-temporal lobe epilepsy is a devastating disease with seizure onset in the hippocampal formation. A fraction of hippocampi samples from epilepsy-surgical procedures reveals a peculiar histological pattern referred to as 'gliosis only' with unresolved pathogenesis and enigmatic sequelae. Here, we hypothesize that 'gliosis only' represents a particular syndrome defined by distinct clinical and molecular characteristics. We curated an in-depth multiparameter integration of systematic clinical, neuropsychological as well as neuropathological analysis from a consecutive cohort of 627 patients, who underwent hippocampectomy for drug-resistant temporal lobe epilepsy. All patients underwent either classic anterior temporal lobectomy or selective amygdalohippocampectomy. On the basis of their neuropathological exam, patients with hippocampus sclerosis and 'gliosis only' were characterized and compared within the whole cohort and within a subset of matched pairs. Integrated transcriptional analysis was performed to address molecular differences between both groups. 'Gliosis only' revealed demographics, clinical and neuropsychological outcome fundamentally different from hippocampus sclerosis. 'Gliosis only' patients had a significantly later seizure onset (16.3 versus 12.2 years, P = 0.005) and worse neuropsychological outcome after surgery compared to patients with hippocampus sclerosis. Epilepsy was less amendable by surgery in 'gliosis only' patients, resulting in a significantly worse rate of seizure freedom after surgery in this subgroup (43% versus 68%, P = 0.0001, odds ratio = 2.8, confidence interval 1.7-4.7). This finding remained significant after multivariate and matched-pairs analysis. The 'gliosis only' group demonstrated pronounced astrogliosis and lack of significant neuronal degeneration in contrast to characteristic segmental neuron loss and fibrillary astrogliosis in hippocampus sclerosis. RNA-sequencing of gliosis only patients deciphered a distinct transcriptional programme that resembles an innate inflammatory response of reactive astrocytes. Our data indicate a new temporal lobe epilepsy syndrome for which we suggest the term 'Innate inflammatory gliosis only'. 'Innate inflammatory gliosis only' is characterized by a diffuse gliosis pattern lacking restricted hippocampal focality and is poorly controllable by surgery. Thus, 'innate inflammatory gliosis only' patients need to be clearly identified by presurgical examination paradigms of pharmacoresistant temporal lobe epilepsy patients; surgical treatment of this subgroup should be considered with great precaution. 'Innate inflammatory gliosis only' requires innovative pharmacotreatment strategies., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2023
- Full Text
- View/download PDF
27. Lactate dehydrogenases promote glioblastoma growth and invasion via a metabolic symbiosis.
- Author
-
Guyon J, Fernandez-Moncada I, Larrieu CM, Bouchez CL, Pagano Zottola AC, Galvis J, Chouleur T, Burban A, Joseph K, Ravi VM, Espedal H, Røsland GV, Daher B, Barre A, Dartigues B, Karkar S, Rudewicz J, Romero-Garmendia I, Klink B, Grützmann K, Derieppe MA, Molinié T, Obad N, Léon C, Seano G, Miletic H, Heiland DH, Marsicano G, Nikolski M, Bjerkvig R, Bikfalvi A, and Daubon T
- Subjects
- Animals, Mice, Lactic Acid, Metabolomics, Lactate Dehydrogenases, Glioblastoma enzymology, Glioblastoma pathology, Brain Neoplasms enzymology, Brain Neoplasms pathology
- Abstract
Lactate is a central metabolite in brain physiology but also contributes to tumor development. Glioblastoma (GB) is the most common and malignant primary brain tumor in adults, recognized by angiogenic and invasive growth, in addition to its altered metabolism. We show herein that lactate fuels GB anaplerosis by replenishing the tricarboxylic acid (TCA) cycle in absence of glucose. Lactate dehydrogenases (LDHA and LDHB), which we found spatially expressed in GB tissues, catalyze the interconversion of pyruvate and lactate. However, ablation of both LDH isoforms, but not only one, led to a reduction in tumor growth and an increase in mouse survival. Comparative transcriptomics and metabolomics revealed metabolic rewiring involving high oxidative phosphorylation (OXPHOS) in the LDHA/B KO group which sensitized tumors to cranial irradiation, thus improving mouse survival. When mice were treated with the antiepileptic drug stiripentol, which targets LDH activity, tumor growth decreased. Our findings unveil the complex metabolic network in which both LDHA and LDHB are integrated and show that the combined inhibition of LDHA and LDHB strongly sensitizes GB to therapy., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
28. Multifunctional mRNA-Based CAR T Cells Display Promising Antitumor Activity Against Glioblastoma.
- Author
-
Meister H, Look T, Roth P, Pascolo S, Sahin U, Lee S, Hale BD, Snijder B, Regli L, Ravi VM, Heiland DH, Sentman CL, Weller M, and Weiss T
- Subjects
- Humans, Mice, Animals, Immunotherapy, Adoptive, NK Cell Lectin-Like Receptor Subfamily K genetics, RNA, Messenger genetics, Xenograft Model Antitumor Assays, Cell Line, Tumor, T-Lymphocytes, Cytokines, Interleukin-12, Tumor Microenvironment genetics, Glioblastoma genetics, Glioblastoma therapy, Glioblastoma pathology, Receptors, Chimeric Antigen genetics
- Abstract
Purpose: Most chimeric antigen receptor (CAR) T-cell strategies against glioblastoma have demonstrated only modest therapeutic activity and are based on persistent gene modification strategies that have limited transgene capacity, long manufacturing processes, and the risk for uncontrollable off-tumor toxicities. mRNA-based T-cell modifications are an emerging safe, rapid, and cost-effective alternative to overcome these challenges, but are underexplored against glioblastoma., Experimental Design: We generated mouse and human mRNA-based multifunctional T cells coexpressing a multitargeting CAR based on the natural killer group 2D (NKG2D) receptor and the proinflammatory cytokines IL12 and IFNα2 and assessed their antiglioma activity in vitro and in vivo., Results: Compared with T cells that either expressed the CAR or cytokines alone, multifunctional CAR T cells demonstrated increased antiglioma activity in vitro and in vivo in three orthotopic immunocompetent mouse glioma models without signs of toxicity. Mechanistically, the coexpression of IL12 and IFNα2 in addition to the CAR promoted a proinflammatory tumor microenvironment and reduced T-cell exhaustion as demonstrated by ex vivo immune phenotyping, cytokine profiling, and RNA sequencing. The translational potential was demonstrated by image-based single-cell analyses of mRNA-modified T cells in patient glioblastoma samples with a complex cellular microenvironment. This revealed strong antiglioma activity of human mRNA-based multifunctional NKG2D CAR T cells coexpressing IL12 and IFNα2 whereas T cells that expressed either the CAR or cytokines alone did not demonstrate comparable antiglioma activity., Conclusions: These data provide a robust rationale for future clinical studies with mRNA-based multifunctional CAR T cells to treat malignant brain tumors., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
29. Spatially resolved multi-omics deciphers bidirectional tumor-host interdependence in glioblastoma.
- Author
-
Ravi VM, Will P, Kueckelhaus J, Sun N, Joseph K, Salié H, Vollmer L, Kuliesiute U, von Ehr J, Benotmane JK, Neidert N, Follo M, Scherer F, Goeldner JM, Behringer SP, Franco P, Khiat M, Zhang J, Hofmann UG, Fung C, Ricklefs FL, Lamszus K, Boerries M, Ku M, Beck J, Sankowski R, Schwabenland M, Prinz M, Schüller U, Killmer S, Bengsch B, Walch AK, Delev D, Schnell O, and Heiland DH
- Subjects
- Humans, Metabolomics methods, Brain Neoplasms pathology, Glioblastoma pathology
- Abstract
Glioblastomas are malignant tumors of the central nervous system hallmarked by subclonal diversity and dynamic adaptation amid developmental hierarchies. The source of dynamic reorganization within the spatial context of these tumors remains elusive. Here, we characterized glioblastomas by spatially resolved transcriptomics, metabolomics, and proteomics. By deciphering regionally shared transcriptional programs across patients, we infer that glioblastoma is organized by spatial segregation of lineage states and adapts to inflammatory and/or metabolic stimuli, reminiscent of the reactive transformation in mature astrocytes. Integration of metabolic imaging and imaging mass cytometry uncovered locoregional tumor-host interdependence, resulting in spatially exclusive adaptive transcriptional programs. Inferring copy-number alterations emphasizes a spatially cohesive organization of subclones associated with reactive transcriptional programs, confirming that environmental stress gives rise to selection pressure. A model of glioblastoma stem cells implanted into human and rodent neocortical tissue mimicking various environments confirmed that transcriptional states originate from dynamic adaptation to various environments., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
30. T-cell dysfunction in the glioblastoma microenvironment is mediated by myeloid cells releasing interleukin-10.
- Author
-
Ravi VM, Neidert N, Will P, Joseph K, Maier JP, Kückelhaus J, Vollmer L, Goeldner JM, Behringer SP, Scherer F, Boerries M, Follo M, Weiss T, Delev D, Kernbach J, Franco P, Schallner N, Dierks C, Carro MS, Hofmann UG, Fung C, Sankowski R, Prinz M, Beck J, Salié H, Bengsch B, Schnell O, and Heiland DH
- Subjects
- Adult, Aged, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Cell Communication immunology, Cell Line, Tumor, Female, Glioblastoma drug therapy, Glioblastoma pathology, Healthy Volunteers, Heme Oxygenase-1 metabolism, Humans, Immunotherapy methods, Janus Kinase Inhibitors pharmacology, Janus Kinase Inhibitors therapeutic use, Janus Kinases antagonists & inhibitors, Janus Kinases metabolism, Male, Middle Aged, Neocortex cytology, Neocortex immunology, Neocortex pathology, Primary Cell Culture, RNA-Seq, STAT Transcription Factors metabolism, Signal Transduction drug effects, Signal Transduction immunology, Single-Cell Analysis, T-Lymphocytes drug effects, T-Lymphocytes metabolism, Tissue Culture Techniques, Tumor Escape, Tumor Microenvironment immunology, Brain Neoplasms immunology, Glioblastoma immunology, Interleukin-10 metabolism, Myeloid Cells metabolism, T-Lymphocytes immunology
- Abstract
Despite recent advances in cancer immunotherapy, certain tumor types, such as Glioblastomas, are highly resistant due to their tumor microenvironment disabling the anti-tumor immune response. Here we show, by applying an in-silico multidimensional model integrating spatially resolved and single-cell gene expression data of 45,615 immune cells from 12 tumor samples, that a subset of Interleukin-10-releasing HMOX1
+ myeloid cells, spatially localizing to mesenchymal-like tumor regions, drive T-cell exhaustion and thus contribute to the immunosuppressive tumor microenvironment. These findings are validated using a human ex-vivo neocortical glioblastoma model inoculated with patient derived peripheral T-cells to simulate the immune compartment. This model recapitulates the dysfunctional transformation of tumor infiltrating T-cells. Inhibition of the JAK/STAT pathway rescues T-cell functionality both in our model and in-vivo, providing further evidence of IL-10 release being an important driving force of tumor immune escape. Our results thus show that integrative modelling of single cell and spatial transcriptomics data is a valuable tool to interrogate the tumor immune microenvironment and might contribute to the development of successful immunotherapies., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
31. Microglia contribute to the propagation of Aβ into unaffected brain tissue.
- Author
-
d'Errico P, Ziegler-Waldkirch S, Aires V, Hoffmann P, Mezö C, Erny D, Monasor LS, Liebscher S, Ravi VM, Joseph K, Schnell O, Kierdorf K, Staszewski O, Tahirovic S, Prinz M, and Meyer-Luehmann M
- Subjects
- Brain metabolism, Humans, Neurons metabolism, Plaque, Amyloid pathology, Amyloid beta-Peptides metabolism, Microglia metabolism
- Abstract
Microglia appear activated in the vicinity of amyloid beta (Aβ) plaques, but whether microglia contribute to Aβ propagation into unaffected brain regions remains unknown. Using transplantation of wild-type (WT) neurons, we show that Aβ enters WT grafts, and that this is accompanied by microglia infiltration. Manipulation of microglia function reduced Aβ deposition within grafts. Furthermore, in vivo imaging identified microglia as carriers of Aβ pathology in previously unaffected tissue. Our data thus argue for a hitherto unexplored mechanism of Aβ propagation., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
32. Meclofenamate causes loss of cellular tethering and decoupling of functional networks in glioblastoma.
- Author
-
Schneider M, Vollmer L, Potthoff AL, Ravi VM, Evert BO, Rahman MA, Sarowar S, Kueckelhaus J, Will P, Zurhorst D, Joseph K, Maier JP, Neidert N, d'Errico P, Meyer-Luehmann M, Hofmann UG, Dolf A, Salomoni P, Güresir E, Enger PØ, Chekenya M, Pietsch T, Schuss P, Schnell O, Westhoff MA, Beck J, Vatter H, Waha A, Herrlinger U, and Heiland DH
- Subjects
- Cell Line, Tumor, Cell Proliferation, Humans, In Vitro Techniques, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Meclofenamic Acid pharmacology
- Abstract
Background: Glioblastoma cells assemble to a syncytial communicating network based on tumor microtubes (TMs) as ultra-long membrane protrusions. The relationship between network architecture and transcriptional profile remains poorly investigated. Drugs that interfere with this syncytial connectivity such as meclofenamate (MFA) may be highly attractive for glioblastoma therapy., Methods: In a human neocortical slice model using glioblastoma cell populations of different transcriptional signatures, three-dimensional tumor networks were reconstructed, and TM-based intercellular connectivity was mapped on the basis of two-photon imaging data. MFA was used to modulate morphological and functional connectivity; downstream effects of MFA treatment were investigated by RNA sequencing and fluorescence-activated cell sorting (FACS) analysis., Results: TM-based network morphology strongly differed between the transcriptional cellular subtypes of glioblastoma and was dependent on axon guidance molecule expression. MFA revealed both a functional and morphological demolishment of glioblastoma network architectures which was reflected by a reduction of TM-mediated intercellular cytosolic traffic as well as a breakdown of TM length. RNA sequencing confirmed a downregulation of NCAM and axon guidance molecule signaling upon MFA treatment. Loss of glioblastoma communicating networks was accompanied by a failure in the upregulation of genes that are required for DNA repair in response to temozolomide (TMZ) treatment and culminated in profound treatment response to TMZ-mediated toxicity., Conclusion: The capacity of TM formation reflects transcriptional cellular heterogeneity. MFA effectively demolishes functional and morphological TM-based syncytial network architectures. These findings might pave the way to a clinical implementation of MFA as a TM-targeted therapeutic approach., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
33. Inhibition of metabotropic glutamate receptor III facilitates sensitization to alkylating chemotherapeutics in glioblastoma.
- Author
-
Maier JP, Ravi VM, Kueckelhaus J, Behringer SP, Garrelfs N, Will P, Sun N, von Ehr J, Goeldner JM, Pfeifer D, Follo M, Hannibal L, Walch AK, Hofmann UG, Beck J, Heiland DH, Schnell O, and Joseph K
- Subjects
- Amino Acids pharmacology, Antineoplastic Agents, Alkylating pharmacology, Cell Death drug effects, Cell Line, Tumor, Cell Survival drug effects, Drug Resistance, Neoplasm drug effects, Gene Expression Regulation, Neoplastic drug effects, Glioblastoma genetics, Glioblastoma pathology, Glutamic Acid metabolism, Humans, Kinetics, Neoadjuvant Therapy, Receptors, Metabotropic Glutamate antagonists & inhibitors, Temozolomide pharmacology, Temozolomide therapeutic use, Tumor Microenvironment drug effects, Xanthenes pharmacology, Antineoplastic Agents, Alkylating therapeutic use, Glioblastoma drug therapy, Receptors, Metabotropic Glutamate metabolism
- Abstract
Glioblastoma (GBM), the most malignant tumor of the central nervous system, is marked by its dynamic response to microenvironmental niches. In particular, this cellular plasticity contributes to the development of an immediate resistance during tumor treatment. Novel insights into the developmental trajectory exhibited by GBM show a strong capability to respond to its microenvironment by clonal selection of specific phenotypes. Using the same mechanisms, malignant GBM do develop intrinsic mechanisms to resist chemotherapeutic treatments. This resistance was reported to be sustained by the paracrine and autocrine glutamate signaling via ionotropic and metabotropic receptors. However, the extent to which glutamatergic signaling modulates the chemoresistance and transcriptional profile of the GBM remains unexplored. In this study we aimed to map the manifold effects of glutamate signaling in GBM as the basis to further discover the regulatory role and interactions of specific receptors, within the GBM microenvironment. Our work provides insights into glutamate release dynamics, representing its importance for GBM growth, viability, and migration. Based on newly published multi-omic datasets, we explored the and characterized the functions of different ionotropic and metabotropic glutamate receptors, of which the metabotropic receptor 3 (GRM3) is highlighted through its modulatory role in maintaining the ability of GBM cells to evade standard alkylating chemotherapeutics. We addressed the clinical relevance of GRM3 receptor expression in GBM and provide a proof of concept where we manipulate intrinsic mechanisms of chemoresistance, driving GBM towards chemo-sensitization through GRM3 receptor inhibition. Finally, we validated our findings in our novel human organotypic section-based tumor model, where GBM growth and proliferation was significantly reduced when GRM3 inhibition was combined with temozolomide application. Our findings present a new picture of how glutamate signaling via mGluR3 interacts with the phenotypical GBM transcriptional programs in light of recently published GBM cell-state discoveries., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
34. Astrogliosis Releases Pro-Oncogenic Chitinase 3-Like 1 Causing MAPK Signaling in Glioblastoma.
- Author
-
Wurm J, Behringer SP, Ravi VM, Joseph K, Neidert N, Maier JP, Doria-Medina R, Follo M, Delev D, Pfeifer D, Beck J, Sankowski R, Schnell O, and Heiland DH
- Abstract
Although reactive astrocytes constitute a major component of the cellular environment in glioblastoma, their function and crosstalk to other components of the environment is still poorly understood. Gene expression analysis of purified astrocytes from both the tumor core and non-infiltrated cortex reveals a tumor-related up-regulation of Chitinase 3-like 1 (CHI3L1), a cytokine which is related to inflammation, extracellular tissue remodeling, and fibrosis. Further, we established and validated a co-culture model to investigate the impact of reactive astrocytes within the tumor microenvironment. Here we show that reactive astrocytes promote a subtype-shift of glioblastoma towards the mesenchymal phenotype, driving mitogen-activated protein kinases (MAPK) signaling as well as increased proliferation and migration. In addition, we demonstrate that MAPK signaling is directly caused by a CHI3L1-IL13RA2 co-binding, which leads to increased downstream MAPK and AKT signaling. This novel microenvironmental crosstalk highlights the crucial role of non-neoplastic cells in malignant brain tumors and opens up new perspectives for targeted therapies in glioblastoma.
- Published
- 2019
- Full Text
- View/download PDF
35. Pre-Clinical Testing of Microwave Radiometer and a Pilot Study on the Screening Inflammation of Knee Joints.
- Author
-
Ravi VM, Sharma AK, and Arunachalam K
- Subjects
- Adult, Electromagnetic Phenomena, Female, Hot Temperature, Humans, Male, Middle Aged, Pilot Projects, Radiometry instrumentation, Thermometry instrumentation, Inflammation diagnosis, Knee Joint metabolism, Microwaves
- Abstract
This article presents the pre-clinical evaluation of our custom-built, single-band microwave radiometer centered at 1.3 GHz for deep tissue thermometry, and a pilot study on volunteers for passive detection of inflammation in knee joints. The electromagnetic (EM) compatibility of the battery-operated radiometer for clinical use was assessed as per International Special Committee on Radio Interference (CISPR) 22 standard. The ability to detect inflammation in knee joints was assessed using a substrate integrated waveguide antenna connected to the radiometer. EM compatibility tests carried out in the laboratory indicated device immunity to intentional radiated interference up to -20 dBm injected power in the global system for mobile communication frequency band, and pre-compliance to CISPR 22 standard. Radiometer temperature measurements recorded at the lateral and medial aspects of both knees of 41 volunteers indicated mean temperature greater than 33°C for the diseased sites compared with the mean temperature of 28°C measured for the healthy sites. One-way analysis of variance statistics indicated significantly (P < 0.005) higher radiometer temperature at the diseased sites unlike the healthy sites. Thus, the EM pre-compliance of the device and the potential to measure deep tissue inflammation were demonstrated. Bioelectromagnetics. 2019;40:402-411. © 2019 Bioelectromagnetics Society., (© 2019 Bioelectromagnetics Society.)
- Published
- 2019
- Full Text
- View/download PDF
36. Human organotypic brain slice culture: a novel framework for environmental research in neuro-oncology.
- Author
-
Ravi VM, Joseph K, Wurm J, Behringer S, Garrelfs N, d'Errico P, Naseri Y, Franco P, Meyer-Luehmann M, Sankowski R, Shah MJ, Mader I, Delev D, Follo M, Beck J, Schnell O, Hofmann UG, and Heiland DH
- Subjects
- Adult, Aged, Aged, 80 and over, Astrocytes metabolism, Brain cytology, Brain metabolism, Brain surgery, Cell Movement, Cell Proliferation, Female, Humans, Infant, Male, Middle Aged, Models, Biological, Nerve Tissue cytology, Nerve Tissue metabolism, Nerve Tissue surgery, Temozolomide pharmacology, Tumor Microenvironment, Brain Neoplasms drug therapy, Brain Neoplasms immunology, Brain Neoplasms metabolism, Brain Neoplasms pathology, Glioblastoma drug therapy, Glioblastoma immunology, Glioblastoma metabolism, Glioblastoma pathology, Tissue Culture Techniques methods
- Abstract
When it comes to the human brain, models that closely mimic in vivo conditions are lacking. Living neuronal tissue is the closest representation of the in vivo human brain outside of a living person. Here, we present a method that can be used to maintain therapeutically resected healthy neuronal tissue for prolonged periods without any discernible changes in tissue vitality, evidenced by immunohistochemistry, genetic expression, and electrophysiology. This method was then used to assess glioblastoma (GBM) progression in its natural environment by microinjection of patient-derived tumor cells into cultured sections. The result closely resembles the pattern of de novo tumor growth and invasion, drug therapy response, and cytokine environment. Reactive transformation of astrocytes, as an example of the cellular nonmalignant tumor environment, can be accurately simulated with transcriptional differences similar to those of astrocytes isolated from acute GBM specimens. In a nutshell, we present a simple method to study GBM in its physiological environment, from which valuable insights can be gained. This technique can lead to further advancements in neuroscience, neuro-oncology, and pharmacotherapy., (© 2019 Ravi et al.)
- Published
- 2019
- Full Text
- View/download PDF
37. Tumor-associated reactive astrocytes aid the evolution of immunosuppressive environment in glioblastoma.
- Author
-
Henrik Heiland D, Ravi VM, Behringer SP, Frenking JH, Wurm J, Joseph K, Garrelfs NWC, Strähle J, Heynckes S, Grauvogel J, Franco P, Mader I, Schneider M, Potthoff AL, Delev D, Hofmann UG, Fung C, Beck J, Sankowski R, Prinz M, and Schnell O
- Subjects
- Astrocytes cytology, Brain Neoplasms metabolism, Cell Line, Tumor, Gene Expression Profiling, Humans, Inflammation Mediators, Janus Kinases metabolism, Microglia cytology, Phenotype, STAT Transcription Factors metabolism, Sequence Analysis, RNA, Signal Transduction, Tissue Culture Techniques, Astrocytes metabolism, Cytokines metabolism, Glioblastoma immunology, Microglia metabolism
- Abstract
Reactive astrocytes evolve after brain injury, inflammatory and degenerative diseases, whereby they undergo transcriptomic re-programming. In malignant brain tumors, their function and crosstalk to other components of the environment is poorly understood. Here we report a distinct transcriptional phenotype of reactive astrocytes from glioblastoma linked to JAK/STAT pathway activation. Subsequently, we investigate the origin of astrocytic transformation by a microglia loss-of-function model in a human organotypic slice model with injected tumor cells. RNA-seq based gene expression analysis of astrocytes reveals a distinct astrocytic phenotype caused by the coexistence of microglia and astrocytes in the tumor environment, which leads to a large release of anti-inflammatory cytokines such as TGFβ, IL10 and G-CSF. Inhibition of the JAK/STAT pathway shifts the balance of pro- and anti-inflammatory cytokines towards a pro-inflammatory environment. The complex interaction of astrocytes and microglia cells promotes an immunosuppressive environment, suggesting that tumor-associated astrocytes contribute to anti-inflammatory responses.
- Published
- 2019
- Full Text
- View/download PDF
38. Helical unwinding and side-chain unlocking unravel the outward open conformation of the melibiose transporter.
- Author
-
Wang LY, Ravi VM, Leblanc G, Padrós E, Cladera J, and Perálvarez-Marín A
- Abstract
Molecular dynamics simulations have been used to study the alternate access mechanism of the melibiose transporter from Escherichia coli. Starting from the outward-facing partially occluded form, 2 out of 12 simulations produced an outward full open form and one partially open, whereas the rest yielded fully or partially occluded forms. The shape of the outward-open form resembles other outward-open conformations of secondary transporters. During the transporter opening, conformational changes in some loops are followed by changes in the periplasm region of transmembrane helix 7. Helical curvature relaxation and unlocking of hydrophobic and ionic locks promote the outward opening of the transporter making accessible the substrate binding site. In particular, FRET studies on mutants of conserved aromatic residues of extracellular loop 4 showed lack of substrate binding, emphasizing the importance of this loop for making crucial interactions that control the opening of the periplasmic side. This study indicates that the alternate access mechanism for the melibiose transporter fits better into a flexible gating mechanism rather than the archetypical helical rigid-body rocker-switch mechanism.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.